Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56.015
Filtrar
Mais filtros

Temas
Intervalo de ano de publicação
1.
Cell ; 187(3): 750-763.e20, 2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-38242132

RESUMO

Breastfeeding offers demonstrable benefits to newborns and infants by providing nourishment and immune protection and by shaping the gut commensal microbiota. Although it has been appreciated for decades that breast milk contains complement components, the physiological relevance of complement in breast milk remains undefined. Here, we demonstrate that weanling mice fostered by complement-deficient dams rapidly succumb when exposed to murine pathogen Citrobacter rodentium (CR), whereas pups fostered on complement-containing milk from wild-type dams can tolerate CR challenge. The complement components in breast milk were shown to directly lyse specific members of gram-positive gut commensal microbiota via a C1-dependent, antibody-independent mechanism, resulting in the deposition of the membrane attack complex and subsequent bacterial lysis. By selectively eliminating members of the commensal gut community, complement components from breast milk shape neonate and infant gut microbial composition to be protective against environmental pathogens such as CR.


Assuntos
Proteínas do Sistema Complemento , Microbioma Gastrointestinal , Leite , Animais , Feminino , Humanos , Lactente , Camundongos , Bactérias , Aleitamento Materno , Citrobacter rodentium , Proteínas do Sistema Complemento/análise , Fatores Imunológicos , Saúde do Lactente , Leite Humano , Leite/química , Infecções por Enterobacteriaceae/imunologia
2.
Cell ; 187(19): 5431-5452.e20, 2024 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-39303691

RESUMO

Breastfeeding and microbial colonization during infancy occur within a critical time window for development, and both are thought to influence the risk of respiratory illness. However, the mechanisms underlying the protective effects of breastfeeding and the regulation of microbial colonization are poorly understood. Here, we profiled the nasal and gut microbiomes, breastfeeding characteristics, and maternal milk composition of 2,227 children from the CHILD Cohort Study. We identified robust colonization patterns that, together with milk components, predict preschool asthma and mediate the protective effects of breastfeeding. We found that early cessation of breastfeeding (before 3 months) leads to the premature acquisition of microbial species and functions, including Ruminococcus gnavus and tryptophan biosynthesis, which were previously linked to immune modulation and asthma. Conversely, longer exclusive breastfeeding supports a paced microbial development, protecting against asthma. These findings underscore the importance of extended breastfeeding for respiratory health and highlight potential microbial targets for intervention.


Assuntos
Aleitamento Materno , Leite Humano , Humanos , Feminino , Leite Humano/microbiologia , Lactente , Pré-Escolar , Asma/microbiologia , Asma/prevenção & controle , Asma/imunologia , Microbiota , Microbioma Gastrointestinal , Masculino , Estudos de Coortes , Recém-Nascido
3.
Cell ; 185(26): 4921-4936.e15, 2022 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-36563663

RESUMO

The perinatal period represents a critical window for cognitive and immune system development, promoted by maternal and infant gut microbiomes and their metabolites. Here, we tracked the co-development of microbiomes and metabolomes from late pregnancy to 1 year of age using longitudinal multi-omics data from a cohort of 70 mother-infant dyads. We discovered large-scale mother-to-infant interspecies transfer of mobile genetic elements, frequently involving genes associated with diet-related adaptations. Infant gut metabolomes were less diverse than maternal but featured hundreds of unique metabolites and microbe-metabolite associations not detected in mothers. Metabolomes and serum cytokine signatures of infants who received regular-but not extensively hydrolyzed-formula were distinct from those of exclusively breastfed infants. Taken together, our integrative analysis expands the concept of vertical transmission of the gut microbiome and provides original insights into the development of maternal and infant microbiomes and metabolomes during late pregnancy and early life.


Assuntos
Microbioma Gastrointestinal , Microbiota , Feminino , Humanos , Lactente , Gravidez , Microbioma Gastrointestinal/genética , Microbiota/genética , Mães , Aleitamento Materno , Fezes , Sequências Repetitivas Dispersas
4.
Cell ; 184(15): 3884-3898.e11, 2021 07 22.
Artigo em Inglês | MEDLINE | ID: mdl-34143954

RESUMO

Immune-microbe interactions early in life influence the risk of allergies, asthma, and other inflammatory diseases. Breastfeeding guides healthier immune-microbe relationships by providing nutrients to specialized microbes that in turn benefit the host's immune system. Such bacteria have co-evolved with humans but are now increasingly rare in modern societies. Here we show that a lack of bifidobacteria, and in particular depletion of genes required for human milk oligosaccharide (HMO) utilization from the metagenome, is associated with systemic inflammation and immune dysregulation early in life. In breastfed infants given Bifidobacterium infantis EVC001, which expresses all HMO-utilization genes, intestinal T helper 2 (Th2) and Th17 cytokines were silenced and interferon ß (IFNß) was induced. Fecal water from EVC001-supplemented infants contains abundant indolelactate and B. infantis-derived indole-3-lactic acid (ILA) upregulated immunoregulatory galectin-1 in Th2 and Th17 cells during polarization, providing a functional link between beneficial microbes and immunoregulation during the first months of life.


Assuntos
Bifidobacterium/fisiologia , Sistema Imunitário/crescimento & desenvolvimento , Sistema Imunitário/microbiologia , Antibacterianos/farmacologia , Biomarcadores/metabolismo , Aleitamento Materno , Linfócitos T CD4-Positivos/imunologia , Polaridade Celular , Proliferação de Células , Citocinas/metabolismo , Fezes/química , Fezes/microbiologia , Galectina 1/metabolismo , Microbioma Gastrointestinal , Humanos , Indóis/metabolismo , Recém-Nascido , Inflamação/sangue , Inflamação/genética , Mucosa Intestinal/imunologia , Metaboloma , Leite Humano/química , Oligossacarídeos/metabolismo , Células Th17/imunologia , Células Th2/imunologia , Água
5.
Immunity ; 54(8): 1633-1635, 2021 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-34380062

RESUMO

Immune-system maturation starts early in life, but studies investigating immune-system education in human infants remain scarce. In a recent issue of Cell, Henrick et al. study early gut microbiota and immune-system development in two infant cohorts. The authors describe that Bifidobacteria can use milk sugars to produce immunoregulatory compounds that induce immune tolerance and reduce intestinal inflammation.


Assuntos
Bifidobacterium/metabolismo , Sistema Imunitário/crescimento & desenvolvimento , Intestinos/imunologia , Intestinos/microbiologia , Leite Humano/química , Oligossacarídeos/metabolismo , Animais , Aleitamento Materno , Microbioma Gastrointestinal/fisiologia , Humanos , Tolerância Imunológica/imunologia , Fatores Imunológicos/química , Imunomodulação/imunologia , Lactente , Suécia , Estados Unidos
6.
Nature ; 607(7918): 345-350, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35768512

RESUMO

Enteric viruses like norovirus, rotavirus and astrovirus have long been accepted as spreading in the population through fecal-oral transmission: viruses are shed into feces from one host and enter the oral cavity of another, bypassing salivary glands (SGs) and reaching the intestines to replicate, be shed in feces and repeat the transmission cycle1. Yet there are viruses (for example, rabies) that infect the SGs2,3, making the oral cavity one site of replication and saliva one conduit of transmission. Here we report that enteric viruses productively and persistently infect SGs, reaching titres comparable to those in the intestines. We demonstrate that enteric viruses get released into the saliva, identifying a second route of viral transmission. This is particularly significant for infected infants, whose saliva directly transmits enteric viruses to their mothers' mammary glands through backflow during suckling. This sidesteps the conventional gut-mammary axis route4 and leads to a rapid surge in maternal milk secretory IgA antibodies5,6. Lastly, we show that SG-derived spheroids7 and cell lines8 can replicate and propagate enteric viruses, generating a scalable and manageable system of production. Collectively, our research uncovers a new transmission route for enteric viruses with implications for therapeutics, diagnostics and importantly sanitation measures to prevent spread through saliva.


Assuntos
Saliva , Glândulas Salivares , Viroses , Vírus , Astroviridae , Aleitamento Materno , Células Cultivadas , Fezes/virologia , Feminino , Humanos , Imunoglobulina A/imunologia , Lactente , Norovirus , Rotavirus , Saliva/virologia , Glândulas Salivares/virologia , Esferoides Celulares/virologia , Viroses/transmissão , Viroses/virologia , Vírus/crescimento & desenvolvimento
7.
Immunol Rev ; 326(1): 117-129, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39162048

RESUMO

In this review, we will highlight infants' immune responses to food, emphasizing the unique aspects of early-life immunity and the critical role of breast milk as a food dedicated to infants. Infants are susceptible to inflammatory responses rather than immune tolerance at the mucosal and skin barriers, necessitating strategies to promote oral tolerance that consider this susceptibility. Breast milk provides nutrients for growth and cell metabolism, including immune cells. The content of breast milk, influenced by maternal genetics and environmental exposures, prepares the infant's immune system for the outside world, including solid foods. To do this, breast milk promotes immune system development through antigen-specific and non-antigen-specific immune education by exposing the newborn to food and respiratory allergens and acting on three key targets for food allergy prevention: the gut microbiota, epithelial cells, and immune cells. Building knowledge of how the maternal exposome and human milk composition influence offspring's healthy immune development will lead to recommendations that meet the specific needs of the developing immune system and increase the chances of promoting an appropriate immune response to food in the long term.


Assuntos
Hipersensibilidade Alimentar , Leite Humano , Humanos , Leite Humano/imunologia , Recém-Nascido , Hipersensibilidade Alimentar/imunologia , Animais , Tolerância Imunológica , Alérgenos/imunologia , Microbioma Gastrointestinal/imunologia , Feminino , Sistema Imunitário/imunologia , Sistema Imunitário/metabolismo , Sistema Imunitário/crescimento & desenvolvimento , Alimentos/efeitos adversos , Lactente , Aleitamento Materno
8.
N Engl J Med ; 390(16): 1493-1504, 2024 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-38657245

RESUMO

BACKGROUND: Most moderate-to-late-preterm infants need nutritional support until they are feeding exclusively on their mother's breast milk. Evidence to guide nutrition strategies for these infants is lacking. METHODS: We conducted a multicenter, factorial, randomized trial involving infants born at 32 weeks 0 days' to 35 weeks 6 days' gestation who had intravenous access and whose mothers intended to breast-feed. Each infant was assigned to three interventions or their comparators: intravenous amino acid solution (parenteral nutrition) or dextrose solution until full feeding with milk was established; milk supplement given when maternal milk was insufficient or mother's breast milk exclusively with no supplementation; and taste and smell exposure before gastric-tube feeding or no taste and smell exposure. The primary outcome for the parenteral nutrition and the milk supplement interventions was the body-fat percentage at 4 months of corrected gestational age, and the primary outcome for the taste and smell intervention was the time to full enteral feeding (150 ml per kilogram of body weight per day or exclusive breast-feeding). RESULTS: A total of 532 infants (291 boys [55%]) were included in the trial. The mean (±SD) body-fat percentage at 4 months was similar among the infants who received parenteral nutrition and those who received dextrose solution (26.0±5.4% vs. 26.2±5.2%; adjusted mean difference, -0.20; 95% confidence interval [CI], -1.32 to 0.92; P = 0.72) and among the infants who received milk supplement and those who received mother's breast milk exclusively (26.3±5.3% vs. 25.8±5.4%; adjusted mean difference, 0.65; 95% CI, -0.45 to 1.74; P = 0.25). The time to full enteral feeding was similar among the infants who were exposed to taste and smell and those who were not (5.8±1.5 vs. 5.7±1.9 days; P = 0.59). Secondary outcomes were similar across interventions. Serious adverse events occurred in one infant. CONCLUSIONS: This trial of routine nutrition interventions to support moderate-to-late-preterm infants until full nutrition with mother's breast milk was possible did not show any effects on the time to full enteral feeding or on body composition at 4 months of corrected gestational age. (Funded by the Health Research Council of New Zealand and others; DIAMOND Australian New Zealand Clinical Trials Registry number, ACTRN12616001199404.).


Assuntos
Aleitamento Materno , Nutrição Enteral , Recém-Nascido Prematuro , Nutrição Parenteral , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Aminoácidos/administração & dosagem , Idade Gestacional , Glucose/administração & dosagem , Leite Humano , Olfato , Paladar , Apoio Nutricional , Soluções de Nutrição Parenteral/uso terapêutico , Adiposidade
9.
Trends Immunol ; 44(8): 644-661, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37438187

RESUMO

Childhood allergy, including asthma, eczema, and food allergies, is a major global health burden, with prevalence increasing dramatically and novel interventions needed. Emerging research suggests that human milk oligosaccharides (HMOs), complex glycans found in breastmilk, have allergy-protective properties, indicating exciting therapeutic potential. This review evaluates current literature on the role of HMOs in allergy, assesses underlying immunological mechanisms, and discusses future research needed to translate findings into clinical implications. HMOs may mediate allergy risk through multiple structure-specific mechanisms, including microbiome modification, intestinal barrier maturation, immunomodulation, and gene regulation. Findings emphasize the importance of breastfeeding encouragement and HMO-supplemented formula milk for high allergy-risk infants. Although further investigation is necessary to determine the most efficacious structures against varying allergy phenotypes and their long-term efficacy, HMOs may represent a promising complementary tool for childhood allergy prevention.


Assuntos
Hipersensibilidade Alimentar , Leite Humano , Lactente , Feminino , Humanos , Criança , Fórmulas Infantis/química , Hipersensibilidade Alimentar/prevenção & controle , Aleitamento Materno , Oligossacarídeos/uso terapêutico , Oligossacarídeos/análise
10.
J Immunol ; 212(4): 513-521, 2024 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-38315948

RESUMO

During pregnancy and lactation, the uterus and mammary glands undergo remarkable structural changes to perform their critical reproductive functions before reverting to their original dormant state upon childbirth and weaning, respectively. Underlying this incredible plasticity are complex remodeling processes that rely on coordinated decisions at both the cellular and tissue-subunit levels. With their exceptional versatility, tissue-resident macrophages play a variety of supporting roles in these organs during each stage of development, ranging from maintaining immune homeostasis to facilitating tissue remodeling, although much remains to be discovered about the identity and regulation of individual macrophage subsets. In this study, we review the increasingly appreciated contributions of these immune cells to the reproductive process and speculate on future lines of inquiry. Deepening our understanding of their interactions with the parenchymal or stromal populations in their respective niches may reveal new strategies to ameliorate complications in pregnancy and breastfeeding, thereby improving maternal health and well-being.


Assuntos
Aleitamento Materno , Lactação , Gravidez , Feminino , Humanos , Animais , Lactação/fisiologia , Macrófagos , Desmame , Útero , Glândulas Mamárias Animais/fisiologia
11.
J Immunol ; 213(5): 612-618, 2024 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-39007643

RESUMO

Breast milk confers multiple benefits to the neonate, including passive immunity against multiple microorganisms via Abs. However, it remains unclear whether breast milk-derived Abs affect vaccine-induced immunity in the neonate. We evaluated in C57BL/6 and BALB/c mice whether breastfeeding from an mRNA-SARS-CoV-2-vaccinated dam affects vaccine-induced immunity in neonate mice. Using an experimental model that allows the distinction of maternal Abs and neonate Abs based on their allotype, we show that breastfeeding from an immune dam is associated with reduced vaccine immunity in the neonate. Importantly, mice that breastfed from an immune dam showed reduced numbers of plasma cells after vaccination, relative to mice that breastfed from a naive dam. Our subsequent studies using an mRNA-luciferase reporter system show that passive transfer of Abs through breastfeeding accelerates the clearance of vaccine Ag in suckling mice, resulting in reduced Ag availability. Altogether, maternal Abs transferred through breast milk can protect against infectious microorganisms, but they may also interfere with the neonate's response to vaccination by accelerating the clearance of vaccine Ag. These findings are important for understanding the effects of maternal Abs on the neonate's response to vaccines and may provide insights for improving neonatal vaccines.


Assuntos
Animais Recém-Nascidos , Imunidade Materno-Adquirida , Camundongos Endogâmicos BALB C , Leite Humano , Animais , Camundongos , Feminino , Imunidade Materno-Adquirida/imunologia , Leite Humano/imunologia , Animais Lactentes/imunologia , Camundongos Endogâmicos C57BL , Vacinação , Humanos , Aleitamento Materno
12.
Nature ; 581(7809): 470-474, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32461640

RESUMO

The gut of healthy human neonates is usually devoid of viruses at birth, but quickly becomes colonized, which-in some cases-leads to gastrointestinal disorders1-4. Here we show that the assembly of the viral community in neonates takes place in distinct steps. Fluorescent staining of virus-like particles purified from infant meconium or early stool samples shows few or no particles, but by one month of life particle numbers increase to 109 per gram, and these numbers seem to persist throughout life5-7. We investigated the origin of these viral populations using shotgun metagenomic sequencing of virus-enriched preparations and whole microbial communities, followed by targeted microbiological analyses. Results indicate that, early after birth, pioneer bacteria colonize the infant gut and by one month prophages induced from these bacteria provide the predominant population of virus-like particles. By four months of life, identifiable viruses that replicate in human cells become more prominent. Multiple human viruses were more abundant in stool samples from babies who were exclusively fed on formula milk compared with those fed partially or fully on breast milk, paralleling reports that breast milk can be protective against viral infections8-10. Bacteriophage populations also differed depending on whether or not the infant was breastfed. We show that the colonization of the infant gut is stepwise, first mainly by temperate bacteriophages induced from pioneer bacteria, and later by viruses that replicate in human cells; this second phase is modulated by breastfeeding.


Assuntos
Aleitamento Materno , Trato Gastrointestinal/virologia , Vírus/isolamento & purificação , Adulto , Bacteriólise , Bacteriófagos/genética , Bacteriófagos/isolamento & purificação , Fezes/virologia , Feminino , Microbioma Gastrointestinal , Trato Gastrointestinal/microbiologia , Humanos , Lactente , Recém-Nascido , Lisogenia , Masculino , Mecônio/virologia , Prófagos/genética , Prófagos/isolamento & purificação , Vírus/genética
13.
Nature ; 577(7791): 543-548, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31915378

RESUMO

Although maternal antibodies protect newborn babies from infection1,2, little is known about how protective antibodies are induced without prior pathogen exposure. Here we show that neonatal mice that lack the capacity to produce IgG are protected from infection with the enteric pathogen enterotoxigenic Escherichia coli by maternal natural IgG antibodies against the maternal microbiota when antibodies are delivered either across the placenta or through breast milk. By challenging pups that were fostered by either maternal antibody-sufficient or antibody-deficient dams, we found that IgG derived from breast milk was crucial for protection against mucosal disease induced by enterotoxigenic E. coli. IgG also provides protection against systemic infection by E. coli. Pups used the neonatal Fc receptor to transfer IgG from milk into serum. The maternal commensal microbiota can induce antibodies that recognize antigens expressed by enterotoxigenic E. coli and other Enterobacteriaceae species. Induction of maternal antibodies against a commensal Pantoea species confers protection against enterotoxigenic E. coli in pups. This role of the microbiota in eliciting protective antibodies to a specific neonatal pathogen represents an important host defence mechanism against infection in neonates.


Assuntos
Anticorpos/imunologia , Escherichia coli Enterotoxigênica/imunologia , Infecções por Escherichia coli/imunologia , Infecções por Escherichia coli/prevenção & controle , Imunidade Materno-Adquirida/imunologia , Recém-Nascido/imunologia , Microbiota/imunologia , Leite Humano/imunologia , Animais , Anticorpos/sangue , Anticorpos/metabolismo , Aleitamento Materno , Reações Cruzadas/imunologia , Infecções por Escherichia coli/microbiologia , Feminino , Humanos , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Imunoglobulina G/metabolismo , Masculino , Camundongos , Mães , Pantoea/imunologia , Receptores Fc/imunologia , Receptores Fc/metabolismo , Simbiose/imunologia
14.
Proc Natl Acad Sci U S A ; 120(30): e2221413120, 2023 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-37433002

RESUMO

Effects of micronutrients on brain connectivity are incompletely understood. Analyzing human milk samples across global populations, we identified the carbocyclic sugar myo-inositol as a component that promotes brain development. We determined that it is most abundant in human milk during early lactation when neuronal connections rapidly form in the infant brain. Myo-inositol promoted synapse abundance in human excitatory neurons as well as cultured rat neurons and acted in a dose-dependent manner. Mechanistically, myo-inositol enhanced the ability of neurons to respond to transsynaptic interactions that induce synapses. Effects of myo-inositol in the developing brain were tested in mice, and its dietary supplementation enlarged excitatory postsynaptic sites in the maturing cortex. Utilizing an organotypic slice culture system, we additionally determined that myo-inositol is bioactive in mature brain tissue, and treatment of organotypic slices with this carbocyclic sugar increased the number and size of postsynaptic specializations and excitatory synapse density. This study advances our understanding of the impact of human milk on the infant brain and identifies myo-inositol as a breast milk component that promotes the formation of neuronal connections.


Assuntos
Aleitamento Materno , Leite Humano , Feminino , Lactente , Humanos , Animais , Camundongos , Ratos , Neurônios , Inositol/farmacologia , Açúcares
15.
Lancet ; 403(10433): 1291-1303, 2024 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-38458222

RESUMO

Inflammatory bowel disease (IBD) affects reproductive planning due to psychological effects and mechanical problems related to surgery. Children of people with IBD have an increased risk of about 10% if one parent has IBD and up to 33% if both parents have IBD. The fertility of people with IBD is similar to the general population, but fertility might be reduced in individuals with active IBD, ileal pouch-anal anastomosis, or perianal Crohn's disease. Flaring disease during pregnancy increases complications, such as preterm birth. Thus, disease management with appropriate medications can optimise outcomes. As most medications have minimal fetal risks, people with IBD should be informed about the risks of stopping medications and the importance of maintaining remission. A period of disease remission is advisable before pregnancy and could reduce the risks for both the pregnant person and the fetus. Flexible endoscopy, intestinal ultrasound, and gadolinium-free magnetic resonance enterography are safe during pregnancy. We provide state-of-the-art knowledge on the basis of the latest evidence to ensure successful pregnancy outcomes in controlled IBD.


Assuntos
Doenças Inflamatórias Intestinais , Complicações na Gravidez , Nascimento Prematuro , Gravidez , Feminino , Criança , Humanos , Recém-Nascido , Doenças Inflamatórias Intestinais/tratamento farmacológico , Resultado da Gravidez , Aleitamento Materno , Lactação , Complicações na Gravidez/tratamento farmacológico
16.
Nature ; 572(7767): 112-115, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31308534

RESUMO

Reconstructing the detailed dietary behaviour of extinct hominins is challenging1-particularly for a species such as Australopithecus africanus, which has a highly variable dental morphology that suggests a broad diet2,3. The dietary responses of extinct hominins to seasonal fluctuations in food availability are poorly understood, and nursing behaviours even less so; most of the direct information currently available has been obtained from high-resolution trace-element geochemical analysis of Homo sapiens (both modern and fossil), Homo neanderthalensis4 and living apes5. Here we apply high-resolution trace-element analysis to two A. africanus specimens from Sterkfontein Member 4 (South Africa), dated to 2.6-2.1 million years ago. Elemental signals indicate that A. africanus infants predominantly consumed breast milk for the first year after birth. A cyclical elemental pattern observed following the nursing sequence-comparable to the seasonal dietary signal that is seen in contemporary wild primates and other mammals-indicates irregular food availability. These results are supported by isotopic evidence for a geographical range that was dominated by nutritionally depauperate areas. Cyclical accumulation of lithium in A. africanus teeth also corroborates the idea that their range was characterized by fluctuating resources, and that they possessed physiological adaptations to this instability. This study provides insights into the dietary cycles and ecological behaviours of A. africanus in response to food availability, including the potential cyclical resurgence of milk intake during times of nutritional challenge (as observed in modern wild orangutans5). The geochemical findings for these teeth reinforce the unique place of A. africanus in the fossil record, and indicate dietary stress in specimens that date to shortly before the extinction of Australopithecus in South Africa about two million years ago.


Assuntos
Fósseis , Hominidae , Estações do Ano , Estresse Fisiológico , Dente/química , Animais , Aleitamento Materno , Hominidae/anatomia & histologia , Hominidae/fisiologia , Pongo , Dente/anatomia & histologia , Dente/fisiologia
17.
Proc Natl Acad Sci U S A ; 119(15): e2121720119, 2022 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-35377806

RESUMO

Human breast milk (hBM) is a dynamic fluid that contains millions of cells, but their identities and phenotypic properties are poorly understood. We generated and analyzed single-cell RNA-sequencing (scRNA-seq) data to characterize the transcriptomes of cells from hBM across lactational time from 3 to 632 d postpartum in 15 donors. We found that the majority of cells in hBM are lactocytes, a specialized epithelial subset, and that cell-type frequencies shift over the course of lactation, yielding greater epithelial diversity at later points. Analysis of lactocytes reveals a continuum of cell states characterized by transcriptional changes in hormone-, growth factor-, and milk production-related pathways. Generalized additive models suggest that one subcluster, LC1 epithelial cells, increases as a function of time postpartum, daycare attendance, and the use of hormonal birth control. We identify several subclusters of macrophages in hBM that are enriched for tolerogenic functions, possibly playing a role in protecting the mammary gland during lactation. Our description of the cellular components of breast milk, their association with maternal­infant dyad metadata, and our quantification of alterations at the gene and pathway levels provide a detailed longitudinal picture of hBM cells across lactational time. This work paves the way for future investigations of how a potential division of cellular labor and differential hormone regulation might be leveraged therapeutically to support healthy lactation and potentially aid in milk production.


Assuntos
Lactação , Leite Humano , Aleitamento Materno , Feminino , Perfilação da Expressão Gênica , Humanos , Lactação/genética , Leite Humano/citologia , Leite Humano/metabolismo , RNA-Seq , Transcriptoma
18.
Diabetologia ; 67(10): 2154-2159, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39028360

RESUMO

AIMS/HYPOTHESIS: This study aimed to describe the relationship between breastfeeding episodes and maternal glucose levels, and to assess whether this differs with closed-loop vs open-loop (sensor-augmented pump) insulin therapy. METHODS: Infant-feeding diaries were collected at 6 weeks, 12 weeks and 24 weeks postpartum in a trial of postpartum closed-loop use in 18 women with type 1 diabetes. Continuous glucose monitoring (CGM) data were used to identify maternal glucose patterns within the 3 h of breastfeeding episodes. Generalised mixed models adjusted for breastfeeding episodes in the same woman, repeat breastfeeding episodes, carbohydrate intake, infant age at time of feeding and early pregnancy HbA1c. This was a secondary analysis of data collected during a randomised trial (ClinicalTrials.gov registration no. NCT04420728). RESULTS: CGM glucose remained above 3.9 mmol/l in the 3 h post-breastfeeding for 93% (397/427) of breastfeeding episodes. There was an overall decrease in glucose at nighttime within 3 h of breastfeeding (1.1 mmol l-1 h-1 decrease on average; p=0.009). A decrease in nighttime glucose was observed with open-loop therapy (1.2 ± 0.5 mmol/l) but was blunted with closed-loop therapy (0.4 ± 0.3 mmol/l; p<0.01, open-loop vs closed-loop). CONCLUSIONS/INTERPRETATION: There is a small decrease in glucose after nighttime breastfeeding that usually does not result in maternal hypoglycaemia; this appears to be blunted with the use of closed-loop therapy.


Assuntos
Glicemia , Aleitamento Materno , Diabetes Mellitus Tipo 1 , Sistemas de Infusão de Insulina , Insulina , Período Pós-Parto , Humanos , Feminino , Diabetes Mellitus Tipo 1/tratamento farmacológico , Diabetes Mellitus Tipo 1/sangue , Glicemia/metabolismo , Glicemia/efeitos dos fármacos , Glicemia/análise , Adulto , Insulina/administração & dosagem , Insulina/uso terapêutico , Gravidez , Hipoglicemiantes/administração & dosagem , Hipoglicemiantes/uso terapêutico , Automonitorização da Glicemia , Controle Glicêmico/métodos , Hemoglobinas Glicadas/metabolismo , Hemoglobinas Glicadas/análise , Recém-Nascido , Lactente
19.
Clin Infect Dis ; 78(6): 1629-1631, 2024 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-38185124

RESUMO

To breast feed or not has long been a difficult question for women with human immunodeficiency virus (HIV) in high-income countries, as undetectable HIV in maternal plasma does not translate to zero risk of transmission while breastfeeding, and clean water and formula are readily available. Recent, and more permissive, changes in US and other high-income-country guidelines regarding breastfeeding underscore this issue and acknowledge the information gaps that are essential for informed maternal choice and provider management. These include lack of guidance as to routine monitoring of mothers during lactation, type and length of prophylaxis for infants, and lack of data on factors associated with increased breast-milk viral load and risk of transmission. Ancillary to data are the education and staffing needs for providers participating in the management of breastfeeding individuals. Future studies of breast-milk transmission will need to evaluate these gaps so that we can move transmission to zero.


Assuntos
Aleitamento Materno , Infecções por HIV , Transmissão Vertical de Doenças Infecciosas , Humanos , Infecções por HIV/prevenção & controle , Infecções por HIV/transmissão , Infecções por HIV/epidemiologia , Feminino , Estados Unidos/epidemiologia , Transmissão Vertical de Doenças Infecciosas/prevenção & controle , Leite Humano/virologia , Recém-Nascido , Lactente , Carga Viral
20.
Clin Infect Dis ; 79(1): 202-207, 2024 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-38270916

RESUMO

Given that HIV can be transmitted through breastfeeding, historically, breastfeeding among women with HIV in the US and other resource-rich settings was discouraged. Formula feeding was the mandated feeding option out of concern for breast-milk transmission of HIV, which occurred in 16-24% of cases pre-antiretroviral therapy (pre-ART) use. In January 2023, the US Department of Health and Human Services' Perinatal Guidelines were revised to support shared decision-making for infant feeding choices. Updated clinical trials' data from resource-limited settings suggest the actual breastmilk HIV transmission rate in the context of maternal ART or neonatal postexposure prophylaxis is 0.3-1%. High-income countries are reporting more people with HIV breastfeeding their infants without cases of HIV transmission. We present the reasons for fully embracing breast-/chestfeeding as a viable, safe infant feeding option for HIV-exposed infants in high-income settings, while acknowledging unanswered questions and the need to continually craft more nuanced clinical guidance.


Assuntos
Aleitamento Materno , Infecções por HIV , Transmissão Vertical de Doenças Infecciosas , Humanos , Infecções por HIV/prevenção & controle , Infecções por HIV/tratamento farmacológico , Infecções por HIV/transmissão , Transmissão Vertical de Doenças Infecciosas/prevenção & controle , Feminino , Recém-Nascido , Lactente , Leite Humano/virologia , Países Desenvolvidos , Gravidez
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA